Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where William Hendricks is active.

Publication


Featured researches published by William Hendricks.


Nature Genetics | 2014

Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic mutations in SMARCA4

Pilar Ramos; Anthony N. Karnezis; David Craig; Aleksandar Sekulic; Megan Russell; William Hendricks; Jason J. Corneveaux; Michael T. Barrett; Karey Shumansky; Yidong Yang; Sohrab P. Shah; Leah M Prentice; Marco A. Marra; Jeffrey Kiefer; Victoria Zismann; Bodour Salhia; Jaime Prat; Emanuela D'Angelo; Blaise Clarke; Joseph G. Pressey; John H Farley; Stephen P Anthony; Richard Roden; Heather E. Cunliffe; David Huntsman; Jeffrey M. Trent

Small cell carcinoma of the ovary of hypercalcemic type (SCCOHT) is an extremely rare, aggressive cancer affecting children and young women. We identified germline and somatic inactivating mutations in the SWI/SNF chromatin-remodeling gene SMARCA4 in 69% (9/13) of SCCOHT cases in addition to SMARCA4 protein loss in 82% (14/17) of SCCOHT tumors but in only 0.4% (2/485) of other primary ovarian tumors. These data implicate SMARCA4 in SCCOHT oncogenesis.


The Journal of Pathology | 2016

Dual loss of the SWI/SNF complex ATPases SMARCA4/BRG1 and SMARCA2/BRM is highly sensitive and specific for small cell carcinoma of the ovary, hypercalcaemic type.

Anthony N. Karnezis; Yemin Wang; Pilar Ramos; William Hendricks; Esther Oliva; Emanuela D'Angelo; Jaime Prat; Marisa R. Nucci; Torsten O. Nielsen; Christine Chow; Samuel Leung; Friedrich Kommoss; Stefan Kommoss; Annacarolina da Silva; Brigitte M. Ronnett; Joseph T. Rabban; David Bowtell; Bernard E. Weissman; Jeffrey M. Trent; C. Blake Gilks; David Huntsman

Small cell carcinoma of the ovary, hypercalcaemic type (SCCOHT) is a lethal and sometimes familial ovarian tumour of young women and children. We and others recently discovered that over 90% of SCCOHTs harbour inactivating mutations in the chromatin remodelling gene SMARCA4 with concomitant loss of its encoded protein SMARCA4 (BRG1), one of two mutually exclusive ATPases of the SWI/SNF chromatin remodelling complex. To determine the specificity of SMARCA4 loss for SCCOHT, we examined the expression of SMARCA4 by immunohistochemistry in more than 3000 primary gynaecological tumours. Among ovarian tumours, it was only absent in clear cell carcinoma (15 of 360, 4%). In the uterus, it was absent in endometrial stromal sarcomas (4 of 52, 8%) and high‐grade endometrioid carcinomas (2 of 338, 1%). Recent studies have shown that SMARCA2 (BRM), the other mutually exclusive ATPase of the SWI/SNF complex, is necessary for survival of tumour cells lacking SMARCA4. Therefore, we examined SMARCA2 expression and discovered that all SMARCA4‐negative SCCOHTs also lacked SMARCA2 protein by IHC, including the SCCOHT cell lines BIN67 and SCCOHT1. Among ovarian tumours, the SMARCA4/SMARCA2 dual loss phenotype appears completely specific for SCCOHT. SMARCA2 loss was not due to mutation but rather from an absence of mRNA expression, which was restored by treatment with the histone deacetylase inhibitor trichostatin A. Re‐expression of SMARCA4 or SMARCA2 inhibited the growth of BIN67 and SCCOHT1 cell lines. Our results indicate that SMARCA4 loss, either alone or with SMARCA2, is highly sensitive and specific for SCCOHT and that restoration of either SWI/SNF ATPase can inhibit the growth of SCCOHT cell lines.


Gynecologic Oncology | 2016

The influence of clinical and genetic factors on patient outcome in small cell carcinoma of the ovary, hypercalcemic type

Leora Witkowski; Catherine Goudie; Pilar Ramos; Talia Boshari; Jean Sébastien Brunet; Anthony N. Karnezis; Michel Longy; James A. Knost; Emmanouil Saloustros; W. Glenn McCluggage; Colin J.R. Stewart; William Hendricks; Heather E. Cunliffe; David Huntsman; Patricia Pautier; Douglas A. Levine; Jeffrey M. Trent; Andrew Berchuck; Martin Hasselblatt; William D. Foulkes

OBJECTIVE Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is an aggressive tumor, with long term survival at ~30% in early stage disease. SCCOHT is caused by germline and somatic SMARCA4 mutations, but the effect of the mutation type on patients remains unknown. Furthermore, the rarity of SCCOHT has resulted in varied treatment, with no standardized protocols. We analyzed 293 cases to determine the effect of treatment modalities and SMARCA4 mutations on patient diagnosis and outcome. METHODS In 293 SCCOHT patients we collected information on age and stage at diagnosis, treatment modality (surgery, chemotherapy, radiotherapy, and/or high-dose chemotherapy with autologous stem cell rescue (HDC-aSCR)), SMARCA4 mutation origin (germline/somatic), and overall survival. Cox analysis and log-rank tests were performed on 257 cases with available survival data. RESULTS The strongest prognostic factors were stage at diagnosis (p=2.72e-15) and treatment modality (p=3.87e-13). For FIGO stages II-IV, 5-year survival was 71% for patients who received HDC-aSCR, compared to 25% in patients who received conventional chemotherapy alone following surgery (p=0.002). Patients aged ≥40 had a worse outcome than younger patients (p=0.04). Twenty-six of 60 tested patients carried a germline SMARCA4 mutation, including all patients diagnosed <15years; carriers presented at a younger age than non-carriers (p=0.02). CONCLUSIONS Stage at diagnosis is the most significant prognostic factor in SCCOHT and consolidation with HDC-aSCR may provide the best opportunity for long-term survival. The large fraction of SMARCA4 germline mutations carriers warrants genetic counseling for all patients.


PLOS ONE | 2014

Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials

Melissa Paoloni; Craig P. Webb; Christina Mazcko; David Cherba; William Hendricks; Susan E. Lana; E. J. Ehrhart; Brad Charles; Heather Fehling; Leena Kumar; David M. Vail; Michael Henson; Michael O. Childress; Barbara E. Kitchell; Christopher Kingsley; Seungchan Kim; Mark W. Neff; Barbara Davis; Chand Khanna; Jeffrey M. Trent

Background Molecularly-guided trials (i.e. PMed) now seek to aid clinical decision-making by matching cancer targets with therapeutic options. Progress has been hampered by the lack of cancer models that account for individual-to-individual heterogeneity within and across cancer types. Naturally occurring cancers in pet animals are heterogeneous and thus provide an opportunity to answer questions about these PMed strategies and optimize translation to human patients. In order to realize this opportunity, it is now necessary to demonstrate the feasibility of conducting molecularly-guided analysis of tumors from dogs with naturally occurring cancer in a clinically relevant setting. Methodology A proof-of-concept study was conducted by the Comparative Oncology Trials Consortium (COTC) to determine if tumor collection, prospective molecular profiling, and PMed report generation within 1 week was feasible in dogs. Thirty-one dogs with cancers of varying histologies were enrolled. Twenty-four of 31 samples (77%) successfully met all predefined QA/QC criteria and were analyzed via Affymetrix gene expression profiling. A subsequent bioinformatics workflow transformed genomic data into a personalized drug report. Average turnaround from biopsy to report generation was 116 hours (4.8 days). Unsupervised clustering of canine tumor expression data clustered by cancer type, but supervised clustering of tumors based on the personalized drug report clustered by drug class rather than cancer type. Conclusions Collection and turnaround of high quality canine tumor samples, centralized pathology, analyte generation, array hybridization, and bioinformatic analyses matching gene expression to therapeutic options is achievable in a practical clinical window (<1 week). Clustering data show robust signatures by cancer type but also showed patient-to-patient heterogeneity in drug predictions. This lends further support to the inclusion of a heterogeneous population of dogs with cancer into the preclinical modeling of personalized medicine. Future comparative oncology studies optimizing the delivery of PMed strategies may aid cancer drug development.


Rare diseases (Austin, Tex.) | 2014

Loss of the tumor suppressor SMARCA4 in small cell carcinoma of the ovary, hypercalcemic type (SCCOHT)

Pilar Ramos; Anthony N. Karnezis; William Hendricks; Yemin Wang; Waibhav Tembe; Victoria Zismann; Christophe Legendre; Winnie S. Liang; Megan Russell; David Craig; John H. Farley; Bradley J. Monk; Stephen P. Anthony; Aleksandar Sekulic; Heather E. Cunliffe; David Huntsman; Jeffrey M. Trent

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT), is a rare and understudied cancer with a dismal prognosis. SCCOHTs infrequency has hindered empirical study of its biology and clinical management. However, we and others have recently identified inactivating mutations in the SWI/SNF chromatin remodeling gene SMARCA4 with concomitant loss of SMARCA4 protein in the majority of SCCOHT tumors.1–4 Here we summarize these findings and report SMARCA4 status by targeted sequencing and/or immunohistochemistry (IHC) in an additional 12 SCCOHT tumors, 3 matched germlines, and the cell line SCCOHT-1. We also report the identification of a homozygous inactivating mutation in the gene SMARCB1 in one SCCOHT tumor with wild-type SMARCA4, suggesting that SMARCB1 inactivation may also play a role in the pathogenesis of SCCOHT. To date, SMARCA4 mutations and protein loss have been reported in the majority of 69 SCCOHT cases (including 2 cell lines). These data firmly establish SMARCA4 as a tumor suppressor whose loss promotes the development of SCCOHT, setting the stage for rapid advancement in the biological understanding, diagnosis, and treatment of this rare tumor type.


Journal of the American Chemical Society | 2016

A Pharmacological Chaperone Molecule Induces Cancer Cell Death by Restoring Tertiary DNA Structures in Mutant hTERT Promoters.

Hyun Jin Kang; Yunxi Cui; Holly Yin; Amy Scheid; William Hendricks; Jessica Schmidt; Aleksandar Sekulic; De-Ming Kong; Jeffrey M. Trent; Vijay Gokhale; Hanbin Mao; Laurence H. Hurley

Activation of human telomerase reverse transcriptase (hTERT) is necessary for limitless replication in tumorigenesis. Whereas hTERT is transcriptionally silenced in normal cells, most tumor cells reactivate hTERT expression by alleviating transcriptional repression through diverse genetic and epigenetic mechanisms. Transcription-activating hTERT promoter mutations have been found to occur at high frequencies in multiple cancer types. These mutations have been shown to form new transcription factor binding sites that drive hTERT expression, but this model cannot fully account for differences in wild-type (WT) and mutant promoter activation and has not yet enabled a selective therapeutic strategy. Here, we demonstrate a novel mechanism by which promoter mutations activate hTERT transcription, which also sheds light on a unique therapeutic opportunity. Promoter mutations occur in a core promoter region that forms tertiary structures consisting of a pair of G-quadruplexes involved in transcriptional silencing. We show that promoter mutations exert a detrimental effect on the folding of one of these G-quadruplexes, resulting in a nonfunctional silencer element that alleviates transcriptional repression. We have also identified a small drug-like pharmacological chaperone (pharmacoperone) molecule, GTC365, that acts at an early step in the G-quadruplex folding pathway to redirect mutant promoter G-quadruplex misfolding, partially reinstate the correct folding pathway, and reduce hTERT activity through transcriptional repression. This transcription-mediated repression produces cancer cell death through multiple routes including both induction of apoptosis through inhibition of hTERTs role in regulating apoptosis-related proteins and induction of senescence by decreasing telomerase activity and telomere length. We demonstrate the selective therapeutic potential of this strategy in melanoma cells that overexpress hTERT.


The Journal of Pathology | 2017

The histone methyltransferase EZH2 is a therapeutic target in small cell carcinoma of the ovary, hypercalcemic type

Yemin Wang; Shary Yuting Chen; Anthony N. Karnezis; Shane Colborne; Nancy Dos Santos; Jessica D. Lang; William Hendricks; Krystal A. Orlando; Damian Yap; Friedrich Kommoss; Marcel B. Bally; Gregg B. Morin; Jeffrey M. Trent; Bernard E. Weissman; David Huntsman

Small cell carcinoma of the ovary, hypercalcaemic type (SCCOHT) is a rare but aggressive and untreatable malignancy affecting young women. We and others recently discovered that SMARCA4, a gene encoding the ATPase of the SWI/SNF chromatin‐remodelling complex, is the only gene recurrently mutated in the majority of SCCOHT. The low somatic complexity of SCCOHT genomes and the prominent role of the SWI/SNF chromatin‐remodelling complex in transcriptional control of genes suggest that SCCOHT cells may rely on epigenetic rewiring for oncogenic transformation. Herein, we report that approximately 80% (19/24) of SCCOHT tumour samples have strong expression of the histone methyltransferase EZH2 by immunohistochemistry, with the rest expressing variable amounts of EZH2. Re‐expression of SMARCA4 suppressed the expression of EZH2 in SCCOHT cells. In comparison to other ovarian cell lines, SCCOHT cells displayed hypersensitivity to EZH2 shRNAs and two selective EZH2 inhibitors, GSK126 and EPZ‐6438. EZH2 inhibitors induced cell cycle arrest, apoptosis, and cell differentiation in SCCOHT cells, along with the induction of genes involved in cell cycle regulation, apoptosis, and neuron‐like differentiation. EZH2 inhibitors suppressed tumour growth and improved the survival of mice bearing SCCOHT xenografts. Therefore, our data suggest that loss of SMARCA4 creates a dependency on the catalytic activity of EZH2 in SCCOHT cells and that pharmacological inhibition of EZH2 is a promising therapeutic strategy for treating this disease. Copyright


Genome Research | 2017

Integrated genomic analyses reveal frequent TERT aberrations in acral melanoma.

Winnie S. Liang; William Hendricks; Jeffrey Kiefer; Jessica Schmidt; Shobana Sekar; John D. Carpten; David Craig; Jonathan Adkins; Lori Cuyugan; Zarko Manojlovic; Rebecca F. Halperin; Adrienne Helland; Sara Nasser; Christophe Legendre; Laurence H. Hurley; Karthigayini Sivaprakasam; Douglas B. Johnson; Holly Crandall; Victoria Zismann; Valerie Deluca; Jeeyun Lee; Aleksandar Sekulic; Charlotte E. Ariyan; Jeffrey A. Sosman; Jeffrey M. Trent

Genomic analyses of cutaneous melanoma (CM) have yielded biological and therapeutic insights, but understanding of non-ultraviolet (UV)-derived CMs remains limited. Deeper analysis of acral lentiginous melanoma (ALM), a rare sun-shielded melanoma subtype associated with worse survival than CM, is needed to delineate non-UV oncogenic mechanisms. We thus performed comprehensive genomic and transcriptomic analysis of 34 ALM patients. Unlike CM, somatic alterations were dominated by structural variation and absence of UV-derived mutation signatures. Only 38% of patients demonstrated driver BRAF/NRAS/NF1 mutations. In contrast with CM, we observed PAK1 copy gains in 15% of patients, and somatic TERT translocations, copy gains, and missense and promoter mutations, or germline events, in 41% of patients. We further show that in vitro TERT inhibition has cytotoxic effects on primary ALM cells. These findings provide insight into the role of TERT in ALM tumorigenesis and reveal preliminary evidence that TERT inhibition represents a potential therapeutic strategy in ALM.


Oncotarget | 2017

Analysis of variability in high throughput screening data: applications to melanoma cell lines and drug responses

Kuan Fu Ding; Darren Finlay; Hongwei Yin; William Hendricks; Chris Sereduk; Jeffrey Kiefer; Aleksandar Sekulic; Patricia LoRusso; Kristiina Vuori; Jeffrey M. Trent; Nicholas J. Schork

High-throughput screening (HTS) strategies and protocols have undergone significant development in the last decade. It is now possible to screen hundreds of thousands of compounds, each exploring multiple biological phenotypes and parameters, against various cell lines or model systems in a single setting. However, given the vast amount of data such studies generate, the fact that they use multiple reagents, and are often technician-intensive, questions have been raised about the variability, reliability and reproducibility of HTS results. Assessments of the impact of the multiple factors in HTS studies could arguably lead to more compelling insights into the robustness of the results of a particular screen, as well as the overall quality of the study. We leveraged classical, yet highly flexible, analysis of variance (ANOVA)-based linear models to explore how different factors contribute to the variation observed in a screening study of four different melanoma cell lines and 120 drugs over nine dosages studied in two independent academic laboratories. We find that factors such as plate effects, appropriate dosing ranges, and to a lesser extent, the laboratory performing the screen, are significant predictors of variation in drug responses across the cell lines. Further, we show that when sources of variation are quantified and controlled for, they contextualize claims of inconsistencies and reveal the overall quality of the HTS studies performed at each participating laboratory. In the context of the broader screening study, we show that our analysis can also elucidate the robust effects of drugs, even those within specific cell lines.


PLOS Genetics | 2018

Somatic inactivating PTPRJ mutations and dysregulated pathways identified in canine malignant melanoma by integrated comparative genomic analysis

William Hendricks; Victoria Zismann; Karthigayini Sivaprakasam; Christophe Legendre; Kelsey Poorman; Waibhav Tembe; Nieves Perdigones; Jeffrey Kiefer; Winnie S. Liang; Valerie DeLuca; Mitchell S. Stark; Alison L. Ruhe; Roe Froman; Nicholas S. Duesbery; Megan Washington; Jessica Aldrich; Mark W. Neff; Matthew J. Huentelman; Nicholas K. Hayward; Kevin D. Brown; Douglas H. Thamm; Gerald Post; Chand Khanna; Barbara Davis; Matthew Breen; Alexander Sekulic; Jeffrey M. Trent

Canine malignant melanoma, a significant cause of mortality in domestic dogs, is a powerful comparative model for human melanoma, but little is known about its genetic etiology. We mapped the genomic landscape of canine melanoma through multi-platform analysis of 37 tumors (31 mucosal, 3 acral, 2 cutaneous, and 1 uveal) and 17 matching constitutional samples including long- and short-insert whole genome sequencing, RNA sequencing, array comparative genomic hybridization, single nucleotide polymorphism array, and targeted Sanger sequencing analyses. We identified novel predominantly truncating mutations in the putative tumor suppressor gene PTPRJ in 19% of cases. No BRAF mutations were detected, but activating RAS mutations (24% of cases) occurred in conserved hotspots in all cutaneous and acral and 13% of mucosal subtypes. MDM2 amplifications (24%) and TP53 mutations (19%) were mutually exclusive. Additional low-frequency recurrent alterations were observed amidst low point mutation rates, an absence of ultraviolet light mutational signatures, and an abundance of copy number and structural alterations. Mutations that modulate cell proliferation and cell cycle control were common and highlight therapeutic axes such as MEK and MDM2 inhibition. This mutational landscape resembles that seen in BRAF wild-type and sun-shielded human melanoma subtypes. Overall, these data inform biological comparisons between canine and human melanoma while suggesting actionable targets in both species.

Collaboration


Dive into the William Hendricks's collaboration.

Top Co-Authors

Avatar

Jeffrey M. Trent

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

David Huntsman

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Anthony N. Karnezis

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Jeffrey Kiefer

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Pilar Ramos

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Victoria Zismann

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

David Craig

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar

Bernard E. Weissman

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jessica D. Lang

Translational Genomics Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge